Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Nontoxic antimicrobials that evade drug resistance

Abstract

Drugs that act more promiscuously provide fewer routes for the emergence of resistant mutants. This benefit, however, often comes at the cost of serious off-target and dose-limiting toxicities. The classic example is the antifungal amphotericin B (AmB), which has evaded resistance for more than half a century. We report markedly less toxic amphotericins that nevertheless evade resistance. They are scalably accessed in just three steps from the natural product, and they bind their target (the fungal sterol ergosterol) with far greater selectivity than AmB. Hence, they are less toxic and far more effective in a mouse model of systemic candidiasis. To our surprise, exhaustive efforts to select for mutants resistant to these more selective compounds revealed that they are just as impervious to resistance as AmB. Thus, highly selective cytocidal action and the evasion of resistance are not mutually exclusive, suggesting practical routes to the discovery of less toxic, resistance-evasive therapies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Synthesis of AmB urea derivatives.
Figure 2: Sterol extraction and binding capacities of AmB ureas.
Figure 3: Efficacy and toxicity of AmB ureas in mice.
Figure 4: Characterization of mechanisms and costs of resistance to AmB ureas.

Similar content being viewed by others

References

  1. Li, J. et al. Colistin: the re-emerging antibiotic for multidrug-resistant Gram-negative bacterial infections. Lancet Infect. Dis. 6, 589–601 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Cortes, J.E. et al. A phase 2 trial of ponatinib in Philadelphia chromosome–positive leukemias. N. Engl. J. Med. 369, 1783–1796 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Ellis, D. Amphotericin B: spectrum and resistance. J. Antimicrob. Chemother. 49, 7–10 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Brown, G. D. et al. Hidden killers: human fungal infections. Sci. Transl. Med. 4, 165rv13 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Volmer, A.A., Szpilman, A.M. & Carreira, E.M. Synthesis and biological evaluation of amphotericin B derivatives. Nat. Prod. Rep. 27, 1329–1349 (2010).

    Article  CAS  PubMed  Google Scholar 

  6. Ermishkin, L.N., Kasumov, K.M. & Potzeluyev, V.M. Single ionic channels induced in lipid bilayers by polyene antibiotics amphotericin B and nystatine. Nature 262, 698–699 (1976).

    Article  CAS  PubMed  Google Scholar 

  7. Gray, K.C. et al. Amphotericin primarily kills yeast by simply binding ergosterol. Proc. Natl. Acad. Sci. USA 109, 2234–2239 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Anderson, T.M. et al. Amphotericin forms an extramembranous and fungicidal sterol sponge. Nat. Chem. Biol. 10, 400–406 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wilcock, B.C., Endo, M.M., Uno, B.E. & Burke, M.D. C2′-OH of amphotericin B plays an important role in binding the primary sterol of human cells but not yeast cells. J. Am. Chem. Soc. 135, 8488–8491 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhang, Y.-Q. et al. Requirement for ergosterol in V-ATPase function underlies antifungal activity of azole drugs. PLoS Pathog. 6, e1000939 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Heese-Peck, A. et al. Multiple functions of sterols in yeast endocytosis. Mol. Biol. Cell 13, 2664–2680 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kato, M. & Wickner, W. Ergosterol is required for the Sec18/ATP-dependent priming step of homotypic vacuole fusion. EMBO J. 20, 4035–4040 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Klose, C. et al. Yeast lipids can phase-separate into micrometer-scale membrane domains. J. Biol. Chem. 285, 30224–30232 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jin, H., McCaffery, J.M. & Grote, E. Ergosterol promotes pheromone signaling and plasma membrane fusion in mating yeast. J. Cell Biol. 180, 813–826 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Vincent, B.M., Lancaster, A.K., Scherz-Shouval, R., Whitesell, L. & Lindquist, S. Fitness trade-offs restrict the evolution of resistance to amphotericin B. PLoS Biol. 11, e1001692 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Palacios, D.S., Dailey, I., Siebert, D.M., Wilcock, B.C. & Burke, M.D. Synthesis-enabled functional group deletions reveal key underpinnings of amphotericin B ion channel and antifungal activities. Proc. Natl. Acad. Sci. USA 108, 6733–6738 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Neumann, A., Baginski, M. & Czub, J. How do sterols determine the antifungal activity of amphotericin B? Free energy of binding between the drug and its membrane targets. J. Am. Chem. Soc. 132, 18266–18272 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Matsumori, N., Sawada, Y. & Murata, M. Mycosamine orientation of amphotericin B controlling interaction with ergosterol: sterol-dependent activity of conformation-restricted derivatives with an amino-carbonyl bridge. J. Am. Chem. Soc. 127, 10667–10675 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Jarzembska, K.N. et al. Controlled crystallization, structure, and molecular properties of iodoacetylamphotericin B. Cryst. Growth Des. 12, 2336–2345 (2012).

    Article  CAS  Google Scholar 

  20. Neant-Fery, M. et al. Molecular basis for the thiol sensitivity of insulin-degrading enzyme. Proc. Natl. Acad. Sci. USA 105, 9582–9587 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Duggan, K.C. et al. (R)-Profens are substrate-selective inhibitors of endocannabinoid oxygenation by COX-2. Nat. Chem. Biol. 7, 803–809 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. MacPherson, D.T. et al. in Recent Advances in the Chemistry of Anti-infective Agents Vol. 119, 205–222 (Royal Society of Chemistry, 1993).

  23. Power, P. et al. Engineered synthesis of 7-oxo- and 15-deoxy-15-oxo-amphotericins: insights into structure-activity relationships in polyene antibiotics. Chem. Biol. 15, 78–86 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Carmody, M. et al. Biosynthesis of amphotericin derivatives lacking exocyclic carboxyl groups. J. Biol. Chem. 280, 34420–34426 (2005).

    Article  CAS  PubMed  Google Scholar 

  25. Byrne, B., Carmody, M., Gibson, E., Rawlings, B. & Caffrey, P. Biosynthesis of deoxyamphotericins and deoxyamphoteronolides by engineered strains of Streptomyces nodosus. Chem. Biol. 10, 1215–1224 (2003).

    Article  CAS  PubMed  Google Scholar 

  26. Maeda, H., Suzuki, M., Sugano, H. & Matsumoto, K. A facile synthesis of (S)-isoserine from (S)-malic acid. Synthesis 1988, 401–402 (1988).

    Article  Google Scholar 

  27. Palacios, D.S., Anderson, T.M. & Burke, M.D.A. Post-PKS oxidation of the amphotericin B skeleton predicted to be critical for channel formation is not required for potent antifungal activity. J. Am. Chem. Soc. 129, 13804–13805 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Bonner, D.P., Mechlinski, W. & Schaffner, C.P. Polyene macrolide derivatives. 3. Biological properties of polyene macrolide ester salts. J. Antibiot. 25, 261–262 (1972).

    Article  CAS  Google Scholar 

  29. Keim, G.R. et al. Comparative toxicological studies of amphotericin B methyl ester and amphotericin B in mice, rats, and dogs. Antimicrob. Agents Chemother. 10, 687–690 (1976).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tevyashova, A.N. et al. Structure-antifungal activity relationships of polyene antibiotics of the amphotericin B group. Antimicrob. Agents Chemother. 57, 3815–3822 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Paquet, V., Volmer, A.A. & Carreira, E.M. Synthesis and in vitro biological properties of novel cationic derivatives of amphotericin B. Chemistry 14, 2465–2481 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Pfaller, M. et al. Epidemiology and outcomes of candidemia in 3648 patients: data from the Prospective Antifungal Therapy (PATH Alliance®) registry, 2004–2008. Diagn. Microbiol. Infect. Dis. 74, 323–331 (2012).

    Article  PubMed  Google Scholar 

  33. Cruz, M.C. et al. Immunosuppressive and nonimmunosuppressive cyclosporine analogs are toxic to the opportunistic fungal pathogen Cryptococcus neoformans via cyclophilin-dependent inhibition of calcineurin. Antimicrob. Agents Chemother. 44, 143–149 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lepak, A.J., Marchillo, K., VanHecker, J. & Andes, D.R. Posaconazole pharmacodynamic target determination against wild-type and Cyp51 mutant isolates of Aspergillus fumigatus in an in vivo model of invasive pulmonary aspergillosis. Antimicrob. Agents Chemother. 57, 579–585 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ellis, J.K. et al. Metabolic response to low-level toxicant exposure in a novel renal tubule epithelial cell system. Mol. Biosyst. 7, 247–257 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Zager, R.A. Polyene antibiotics: relative degrees of in vitro cytotoxicity and potential effects on tubule phospholipid and ceramide content. Am. J. Kidney Dis. 36, 238–249 (2000).

    Article  CAS  PubMed  Google Scholar 

  37. Andes, D., Stamsted, T. & Conklin, R. Pharmacodynamics of amphotericin B in a neutropenic-mouse disseminated-candidiasis model. Antimicrob. Agents Chemother. 45, 922–926 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Pfaller, M.A. Antifungal drug resistance: mechanisms, epidemiology, and consequences for treatment. Am. J. Med. 125, S3–S13 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Sanglard, D., Ischer, F., Parkinson, T., Falconer, D. & Bille, J. Candida albicans mutations in the ergosterol biosynthetic pathway and resistance to several antifungal agents. Antimicrob. Agents Chemother. 47, 2404–2412 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Pfaller, M.A. et al. Wild-type MIC distributions and epidemiological cutoff values for amphotericin B, flucytosine, and itraconazole and Candida spp. as determined by CLSI broth microdilution. J. Clin. Microbiol. 50, 2040–2046 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Pinto, J.P., Machado, R., Xavier, J.G. & Futschik, M.E. Targeting molecular networks for drug research. Front. Genet. 5, 160 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Grongsaard, P. et al. Convergent, kilogram scale synthesis of an Akt kinase inhibitor. Org. Process Res. Dev. 16, 1069–1081 (2012).

    Article  CAS  Google Scholar 

  43. Dailey, I. Synthesis and Function of the Conserved Motif of Mycosamine Containing Polyene Macrolides. PhD thesis,University of Illinois at Urbana-Champaign, (2012).

  44. Kotler-Brajtburg, J. et al. Classification of polyene antibiotics according to chemical structure and biological effects. Antimicrob. Agents Chemother. 15, 716–722 (1979).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Moonis, M., Ahmad, I. & Bachhawat, B.K. Liposomal hamycin in the control of experimental aspergillosis in mice—relative toxicity, therapeutic efficacy and tissue distribution of free and liposomal hamycin. Indian J. Biochem. Biophys. 29, 339–345 (1992).

    CAS  PubMed  Google Scholar 

  46. Hasper, H.E. et al. An alternative bactericidal mechanism of action for lantibiotic peptides that target lipid II. Science 313, 1636–1637 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Ling, L.L. et al. A new antibiotic kills pathogens without detectable resistance. Nature 517, 455–459 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Schneider, T. et al. Plectasin, a fungal defensin, targets the bacterial cell wall precursor lipid II. Science 328, 1168–1172 (2010).

    Article  CAS  PubMed  Google Scholar 

  49. Laganowsky, A. et al. Membrane proteins bind lipids selectively to modulate their structure and function. Nature 510, 172–175 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Han, X., Yang, K. & Gross, R.W. Multi-dimensional mass spectrometry-based shotgun lipidomics and novel strategies for lipidomic analyses. Mass Spectrom. Rev. 31, 134–178 (2012).

    Article  CAS  PubMed  Google Scholar 

  51. Pangborn, A.B., Giardello, M.A., Grubbs, R.H., Rosen, R.K. & Timmers, F.J. Safe and convenient procedure for solvent purification. Organometallics 15, 1518–1520 (1996).

    Article  CAS  Google Scholar 

  52. Clinical Laboratory Standards Institute (CLSI). Reference method for broth dilution antifungal susceptibility testing of yeasts; approved standard, 3rd ed., CLSI document M27–A3 (Clinical and Laboratory Standards Institute, Wayne, PA, 2008).

  53. Clinical Laboratory Standards Institute (CLSI). Reference method for broth dilution antifungal susceptibility testing of filamentous fungi; approved standard, 2nd edn. CLSI document M38–A2 (Clinical Laboratory Standards Institute, Wayne, PA, 2007).

  54. Sebaugh, J.L. Guidelines for accurate EC50/IC50 estimation. Pharm. Stat. 10, 128–134 (2011).

    Article  CAS  PubMed  Google Scholar 

  55. Andes, D. & van Ogtrop, M. Characterization and quantitation of the pharmacodynamics of fluconazole in a neutropenic murine disseminated candidiasis infection model. Antimicrob. Agents Chemother. 43, 2116–2120 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Andes, D. & van Ogtrop, M. In vivo characterization of the pharmacodynamics of flucytosine in a neutropenic murine disseminated candidiasis model. Antimicrob. Agents Chemother. 44, 938–942 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Portions of this work were supported by the US National Institutes of Health (R01GM080436, R01GM080436-S), the Howard Hughes Medical Institute (HHMI) and the Mathers Foundation. M.D.B. is an HHMI Early Career Scientist, and S.L. is an HHMI Investigator.

Author information

Authors and Affiliations

Authors

Contributions

S.A.D. and M.D.B. conceived the study and oversaw design of synthesis, biophysical and several biological experiments. B.M.V., L.W. and S.L. designed resistance studies. D.R.A. designed mouse toxicity and efficacy studies. S.A.D. synthesized all of the compounds. B.M.V. executed all of the resistance studies. M.M.E. performed sterol binding and designed and performed cell toxicity assays. K.M. performed efficacy and toxicity studies in mice. S.A.D., B.M.V., S.L. and M.D.B. wrote the manuscript.

Corresponding authors

Correspondence to Susan Lindquist or Martin D Burke.

Ethics declarations

Competing interests

The University of Illinois has filed patents on compounds and chemistry reported herein. These have been licensed to REVOLUTION Medicines, a company for which M.D.B. is a founder.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Table 1, Supplementary Figure 1 and Supplementary Note (PDF 5329 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Davis, S., Vincent, B., Endo, M. et al. Nontoxic antimicrobials that evade drug resistance. Nat Chem Biol 11, 481–487 (2015). https://doi.org/10.1038/nchembio.1821

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.1821

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology